scispace - formally typeset
Open AccessJournal Article

Cancer: A Disease of the Genome

TLDR
The Cancer Genome Atlas (TCGA) is bringing together investigators with a wide range of clinical and basic science skills and asking them to apply these technologies to a full characterization of three cancer types, using a open data access model which will empower many investigators around the world.
Abstract
PL01-01 Decades of research into genetics and molecular biology of cancer are painting an increasingly detailed picture of the molecular basis of the disease, confirming that cancer is clearly a disease of the genome. Some of the genetic factors are hereditary, but most represent somatic mutations acquired during life in tumor suppressor genes, oncogenes, or genes affecting DNA stability. These molecular revelations are increasingly being translated into diagnostic and therapeutic tools. The full characterization of the cancer cell represents our best hope for the ultimate conquest of this disease. The time is now right for applying the increasingly powerful tools of genomics, arising from the Human Genome Project, to achieve a comprehensive molecular characterization of cancer. Accordingly, the National Cancer Institute and the National Human Genome Research Institute have joined together to initiate a bold new program - The Cancer Genome Atlas (TCGA). Beginning as a three-year pilot project, TCGA is bringing together investigators with a wide range of clinical and basic science skills and asking them to apply these technologies to a full characterization of three cancer types, using a open data access model which will empower many investigators around the world (see http://www.cancergenome.nih.gov). The components of TCGA include 1) a Biospecimen Repository where tumor specimens will be characterized and biomolecules prepared for analysis; 2) a sequencing of tumor DNA (and control DNA from the same individual), to be conducted by three DNA sequencing centers, focused initially on a candidate gene list appropriate for the particular tumor at hand, but ultimately aiming to extend this list to include the entire genome; and 3) a series of seven Cancer Genome Characterization Centers (CGCCs) that will apply a wide variety of high throughput technologies to look at copy number changes, gene expression and epigenetic alterations in the tumors. All of the data will be quality-checked and made rapidly accessible to the scientific community, through the caBIG database supported by the National Cancer Institute. The initial tumors chosen for study are glioblastoma multiforme, squamous cell lung cancer, and ovarian cancer. Approximately 500 tumors of each type will be studied during the three year pilot period. A “pre-pilot” project supported by NHGRI has been conducting similar analyses on adenocarcinoma of the lung and has already uncovered important findings based upon this kind of comprehensive genomic analysis. This experience adds further confidence that the coordinated, multi-disciplinary, technologically advanced and open data access approach to highly characterized tumors will yield many new findings. Uncovering the genomic roots of cancer will provide many new insights into subtypes of disease that are currently lumped together, allowing more precise correlations with prognosis and response to therapy. Perhaps most significantly, these studies are expected to uncover new drug targets that will catalyze the next generation of cancer therapies that will be much more specific and individualized than many current standard approaches. One can therefore anticipate major changes in the approach to diagnosis, prevention and treatment of cancer in the coming years.

read more

Citations
More filters
Journal ArticleDOI

A future of cancer prevention and cures: highlights of the Centennial Meeting of the American Association for Cancer Research

TL;DR: Advances in the clinical application of genomics, epigenomics and proteomics to diagnose, monitor and prognosticate cancer development led to a dramatic increase in the number of presentations with a translational focus at this year's AACR meeting.
Journal ArticleDOI

Racial and ethnic disparities in a real-world precision oncology data registry

TL;DR: In this article , an analysis of Project Genomics Evidence Neoplasia Information Exchange (GENIE), a real-world cancer data registry designed to accelerate precision oncology discovery, indicates that minorities do not have sufficient representation, which may impact the validity of studies directly comparing mutational profiles between racial/ethnic groups.

Using Machine Learning to Predict Chemotherapy Response in Cell Lines and Patients Based on Genetic Expression

Dimo Angelov
TL;DR: It was concluded that binary classification was the best method for training models due to the limited size of available cell line data and Sequential backward feature selection was able to select genes for the models that produced good results, however the greedy algorithm has limitations.
Related Papers (5)
Trending Questions (1)
Cancer as a Multi-Step Genetic Disease:

Cancer is a multi-step genetic disease caused by somatic mutations acquired during life in tumor suppressor genes, oncogenes, or genes affecting DNA stability.